Skip to main content

A Scalable Lentiviral Vector Production and Purification Method Using Mustang Q Chromatography and Tangential Flow Filtration

  • Protocol
  • First Online:
Viral Vectors for Gene Therapy

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1937))

Abstract

Lentiviral vectors have rapidly become a favorite tool for research and clinical gene transfer applications which seek to permanently introduce alterations in the genome. This status can be attributed primarily to their ability to transduce dividing as well as quiescent cells. When coupled with internal promotor selection to drive expression in one cell type but not another, the ease with which the vectors can be pseudotyped to either restrict or expand tropism offers unique opportunities previously unavailable to the researcher to manipulate the genome. Although LV can be produced from stable packaging cell lines and/or in suspension culture, by and far, most LV vectors are produced using adherent 293 T cells grown in plasticware and production plasmids transiently transfected with either PEI or Calcium Phosphate. The media is usually changed and un-concentrated vector supernatant collected between 24 and 48 h post-transfection. The supernatant may then be purified by Mustang Q chromatography, concentrated by Tangential Flow Filtration, and finally diafiltered into the final formulation buffer of choice. Here we describe a pilot scale method for the manufacture of a Lentiviral vector that purifies and concentrates approximately 6 L of un-concentrated LV supernatant to approximately 150 mL. Typical titers for most vector constructs range between 1 × 108 and 1 × 109 infectious particles per mL. This method may be performed reiteratively to increase total volume or can be further scaled up to increase yield.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 189.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Otto-Wilhelm M, Hebben M, Bovolenta C (2016) Production of lentiviral vectors. Mol Ther Methods Clin Dev 3:16017. https://doi.org/10.1038/mtm.2016.17

    Article  CAS  Google Scholar 

  2. Coffin J, Hughes S, Varmus H (eds) (1997) Retroviruses. Cold Spring Harbor Laboratory Press, New York

    Google Scholar 

  3. Rosenberg S, Aebersold P, Cornetta K et al (1990) Gene transfer into humans--immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med 323:570–578

    Article  CAS  PubMed  Google Scholar 

  4. Blaese R, Culver K, Miller A et al (1995) T lymphocyte-directed gene therapy for ADA− SCID: initial trial results after 4 years. Science 270:475–480

    Article  CAS  PubMed  Google Scholar 

  5. Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G et al (2000) Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288:669–672

    Article  CAS  PubMed  Google Scholar 

  6. Kohn D, Weinberg K, Nolta J et al (1995) Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nat Med 1:1017–1023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Aiuti A, Slavin S, Aker M et al (2002) Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296:2410–2413. https://doi.org/10.1126/science.1070104

    Article  CAS  PubMed  Google Scholar 

  8. Hacein-Bey-Abina S, Von Kalle C, Schmidt M et al (2003) LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302:415–419. https://doi.org/10.1126/science.1088547

    Article  CAS  PubMed  Google Scholar 

  9. Bushman F (2007) Retroviral integration and human gene therapy. J Clin Invest 117(8):2083–2086

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Schröder A, Shinn P, Chen H et al (2002) HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110(4):521–529

    Article  PubMed  Google Scholar 

  11. Mitchell R, Beitzel B, Schroder A et al (2004) Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol 2(8):E234. https://doi.org/10.1371/journal.pbio.0020234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schambach A, Swaney W, van der Loo J (2009) Design and production of retro- and lentiviral vectors for gene expression in hematopoietic cells. Methods Mol Biol 506:191–205. https://doi.org/10.1007/978-1-59745-409-4_14

    Article  CAS  PubMed  Google Scholar 

  13. Modlich U, Bohne J, Schmidt M et al (2006) Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity. Blood 108:2545–2553. https://doi.org/10.1182/blood-2005-08-024976

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dull T, Zufferey R, Kelly M et al (1998) A third-generation lentivirus vector with a conditional packaging system. J Virol 72(11):8463–8471

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gama-Norton L, Botezatu L, Herrmann S et al (2011) Lentivirus production is influenced by SV40 large T-antigen and chromosomal integration of the vector in HEK293 cells. Hum Gene Ther 22:1269–1279. https://doi.org/10.1089/hum.2010.143

    Article  CAS  PubMed  Google Scholar 

  16. Slepushkin V, Chang N, Cohen R et al (2003) Large-scale purification of a lentiviral vector by size exclusion chromatography or Mustang Q ion exchange chromatography. Bioprocess J 2:89–95

    Article  Google Scholar 

  17. Leath A, Cornetta K (2012) Developing novel lentiviral vectors into clinical products. Meth Enzymol 507:89–108. https://doi.org/10.1016/B978-0-12-386509-0.00005-3

    Article  CAS  Google Scholar 

Download references

Acknowledgments

Our facility is generously supported by the Cincinnati Children’s Hospital Research Foundation. Most importantly, we would like to acknowledge the hard work and dedication of the entire staff of the Translational Core Laboratories at Cincinnati Children’s Hospital; without it, our contributions to the gene therapy field would not be possible.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Scott R. Witting .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Tinch, S., Szczur, K., Swaney, W., Reeves, L., Witting, S.R. (2019). A Scalable Lentiviral Vector Production and Purification Method Using Mustang Q Chromatography and Tangential Flow Filtration. In: Manfredsson, F., Benskey, M. (eds) Viral Vectors for Gene Therapy. Methods in Molecular Biology, vol 1937. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-9065-8_8

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-9065-8_8

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-9064-1

  • Online ISBN: 978-1-4939-9065-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics